Full Text:  <902>

Suppl. Mater.: 

Summary:  <245>

CLC number: 

On-line Access: 2023-01-10

Received: 2022-05-10

Revision Accepted: 2022-07-28

Crosschecked: 2023-01-16

Cited: 0

Clicked: 1019

Citations:  Bibtex RefMan EndNote GB/T7714

 ORCID:

Lin ZHANG

https://orcid.org/0000-0001-8567-0238

Dianshuai GAO

Dianshuai GAO, https://orcid.org/0000-0001-8567-0238

-   Go to

Article info.
Open peer comments

Journal of Zhejiang University SCIENCE B

Accepted manuscript available online (unedited version)


Hypoxia-induced ROS aggravate tumor progression through HIF-1α-SERPINE1 signaling in glioblastoma


Author(s):  Lin ZHANG, Yuanyuan CAO, Xiaoxiao GUO, Xiaoyu WANG, Xiao HAN, Kouminin KANWORE, Xiaoliang HONG, Han ZHOU, Dianshuai GAO

Affiliation(s):  School of Nursing, Xuzhou Medical University, Xuzhou 221004, China; more

Corresponding email(s):  gds@xzhmu.edu.cn

Key Words:  Glioblastoma; Hypoxia; Reactive oxygen species (ROS); Hypoxia-inducible factor-1α (HIF-1α); Serine protease inhibitor family E member 1 (SERPINE1)


Share this article to: More <<< Previous Paper|Next Paper >>>

Lin ZHANG, Yuanyuan CAO, Xiaoxiao GUO, Xiaoyu WANG, Xiao HAN, Kouminin KANWORE, Xiaoliang HONG, Han ZHOU, Dianshuai GAO. Hypoxia-induced ROS aggravate tumor progression through HIF-1α-SERPINE1 signaling in glioblastoma[J]. Journal of Zhejiang University Science B,in press.Frontiers of Information Technology & Electronic Engineering,in press.https://doi.org/10.1631/jzus.B2200269

@article{title="Hypoxia-induced ROS aggravate tumor progression through HIF-1α-SERPINE1 signaling in glioblastoma",
author="Lin ZHANG, Yuanyuan CAO, Xiaoxiao GUO, Xiaoyu WANG, Xiao HAN, Kouminin KANWORE, Xiaoliang HONG, Han ZHOU, Dianshuai GAO",
journal="Journal of Zhejiang University Science B",
year="in press",
publisher="Zhejiang University Press & Springer",
doi="https://doi.org/10.1631/jzus.B2200269"
}

%0 Journal Article
%T Hypoxia-induced ROS aggravate tumor progression through HIF-1α-SERPINE1 signaling in glioblastoma
%A Lin ZHANG
%A Yuanyuan CAO
%A Xiaoxiao GUO
%A Xiaoyu WANG
%A Xiao HAN
%A Kouminin KANWORE
%A Xiaoliang HONG
%A Han ZHOU
%A Dianshuai GAO
%J Journal of Zhejiang University SCIENCE B
%P 32-49
%@ 1673-1581
%D in press
%I Zhejiang University Press & Springer
doi="https://doi.org/10.1631/jzus.B2200269"

TY - JOUR
T1 - Hypoxia-induced ROS aggravate tumor progression through HIF-1α-SERPINE1 signaling in glioblastoma
A1 - Lin ZHANG
A1 - Yuanyuan CAO
A1 - Xiaoxiao GUO
A1 - Xiaoyu WANG
A1 - Xiao HAN
A1 - Kouminin KANWORE
A1 - Xiaoliang HONG
A1 - Han ZHOU
A1 - Dianshuai GAO
J0 - Journal of Zhejiang University Science B
SP - 32
EP - 49
%@ 1673-1581
Y1 - in press
PB - Zhejiang University Press & Springer
ER -
doi="https://doi.org/10.1631/jzus.B2200269"


Abstract: 
Hypoxia, as an important hallmark of the tumor microenvironment, is a major cause of oxidative stress and plays a central role in various malignant tumors, including glioblastoma. Elevated reactive oxygen species (ROS) in a hypoxic microenvironment promote glioblastoma progression; however, the underlying mechanism has not been clarified. Herein, we found that hypoxia promoted ROS production, and the proliferation, migration, and invasion of glioblastoma cells, while this promotion was restrained by ROS scavengers N-acetyl-L-cysteine (NAC) and diphenyleneiodonium chloride (DPI). Hypoxia-induced ROS activated hypoxia-inducible factor-1α (HIF-1α) signaling, which enhanced cell migration and invasion by epithelial-mesenchymal transition (EMT). Furthermore, the induction of serine protease inhibitor family E member 1 (SERPINE1) was ROS-dependent under hypoxia, and HIF-1α mediated SERPINE1 increase induced by ROS via binding to the SERPINE1 promoter region, thereby facilitating glioblastoma migration and invasion. Taken together, our data revealed that hypoxia-induced ROS reinforce the hypoxic adaptation of glioblastoma by driving the HIF-1α-SERPINE1 signaling pathway, and that targeting ROS may be a promising therapeutic strategy for glioblastoma.

缺氧诱导的ROS通过HIF-1α-SERPINE1信号通路促进胶质母细胞瘤恶性进展

张琳1,2,曹媛媛2,3,郭肖肖2,4,王潇雨2,韩笑5,Kouminin KANWORE2,洪晓亮2,周晗2,高殿帅2
1徐州医科大学护理学院,中国徐州市,221004
2徐州医科大学神经生物学重点实验室,中国徐州市,221004
3泰州市中医院超声科,中国泰州市,225300
4南京医科大学康达学院基础学院,中国连云港市,222000
5南京医科大学第四临床学院,中国南京市,211166
概要:缺氧作为肿瘤微环境的重要特征,是氧化应激的主要原因,在肿瘤(包括胶质母细胞瘤)的恶性进展中发挥重要作用。缺氧微环境中高水平活性氧(ROS)促进胶质母细胞瘤进展的潜在机制尚不清楚。本研究发现缺氧促进了胶质母细胞瘤细胞增殖、迁移和侵袭以及ROS生成,而这种促进作用可被ROS清除剂N-乙酰半胱氨酸(NAC)和二苯基氯化碘盐(DPI)抑制。缺氧诱导的ROS可激活缺氧诱导因子-1α(HIF-1α)信号,通过上皮-间充质转化(EMT)增强细胞迁移和侵袭。此外,在缺氧条件下,HIF-1α可与丝氨酸蛋白酶抑制剂家族E成员1(SERPINE1)启动子区结合,ROS经HIF-1α上调SERPINE1表达,进而促进胶质母细胞瘤细胞迁移和侵袭。综上所述,本研究揭示了缺氧诱导的ROS通过驱动HIF-1α-SERPINE1信号促进胶质母细胞瘤的缺氧适应,靶向ROS可能成为胶质母细胞瘤治疗的一种有效策略。

关键词组:胶质母细胞瘤;缺氧;活性氧(ROS);缺氧诱导因子-1α(HIF-1α);丝氨酸蛋白酶抑制剂家族E成员1(SERPINE1)

Darkslateblue:Affiliate; Royal Blue:Author; Turquoise:Article

Reference

[1]AndrewAS, KleiLR, BarchowskyA, 2001. Nickel requires hypoxia-inducible factor-1α, not redox signaling, to induce plasminogen activator inhibitor-1. Am J Physiol Lung Cell Mol Physiol, 281(3):L607-L615.

[2]AzimiI, PetersenRM, ThompsonEW, et al., 2017. Hypoxia-induced reactive oxygen species mediate N-cadherin and SERPINE1 expression, EGFR signalling and motility in MDA-MB-468 breast cancer cells. Sci Rep, 7:15140.

[3]CarreresL, Mercey-RessejacM, KurmaK, et al., 2022. Chronic intermittent hypoxia increases cell proliferation in hepatocellular carcinoma. Cells, 11(13):2051.

[4]ChandelNS, MaltepeE, GoldwasserE, et al., 1998. Mitochondrial reactive oxygen species trigger hypoxia-induced transcription. Proc Natl Acad Sci USA, 95(20):‍11715-11720.

[5]ChandelNS, McClintockDS, FelicianoCE, et al., 2000. React

[6]ive oxygen species generated at mitochondrial complex III stabilize hypoxia-inducible factor-1α during hypoxia: a mechanism of O2 sensing. J Biol Chem, 275(33):25130-25138.

[7]ChédevilleAL, LourdusamyA, MonteiroAR, et al., 2020. Investigating glioblastoma response to hypoxia. Biomedicines, 8(9):310.

[8]ChenCL, WangSC, LiuP, 2019. Deferoxamine enhanced mitochondrial iron accumulation and promoted cell migration in triple-negative MDA-MB-231 breast cancer cells via a ROS-dependent mechanism. Int J Mol Sci, 20(19):4852.

[9]ChenD, WuYX, QiuYB, et al., 2020. Hyperoside suppresses hypoxia-induced A549 survival and proliferation through ferrous accumulation via AMPK/HO-1 axis. Phytomedicine, 67:153138.

[10]ChenXT, LiZW, YongHM, et al., 2021. Trim21-mediated HIF-1α degradation attenuates aerobic glycolysis to inhibit renal cancer tumorigenesis and metastasis. Cancer Lett, 508:115-126.

[11]ChiuJ, DawesIW, 2012. Redox control of cell proliferation. Trends Cell Biol, 22(11):592-601.

[12]ChuaYL, DufourE, DassaEP, et al., 2010. Stabilization of hypoxia-inducible factor-1α protein in hypoxia occurs independently of mitochondrial reactive oxygen species production. J Biol Chem, 285(41):31277-31284.

[13]DabralS, MueckeC, ValasarajanC, et al., 2019. A RASSF1A-HIF1α loop drives Warburg effect in cancer and pulmonary hypertension. Nat Commun, 10:2130.

[14]Dao TrongP, RöschS, MairbäurlH, et al., 2018. Identification of a prognostic hypoxia-associated gene set in IDH-mutant glioma. Int J Mol Sci, 19(10):2903.

[15]DröseS, 2013. Differential effects of complex II on mitochondrial ROS production and their relation to cardioprotective pre- and postconditioning. Biochim Biophys Acta Bioenerget, 1827(5):578-587.

[16]FandreyJ, Gassmann M, 2009. Oxygen sensing and the activation of the hypoxia inducible factor 1 (HIF-1)‍‒‍Invited Article. In: Gonzalez C, Nurse CA, Peers C (Eds.), Arterial Chemoreceptors. Advances in Experimental Medicine and Biology, Vol. 648. Springer, Dordrecht, p.197-206.

[17]FinkT, KazlauskasA, PoellingerL, et al., 2002. Identification of a tightly regulated hypoxia-response element in the promoter of human plasminogen activator inhibitor-1. Blood, 99(6):2077-2083.

[18]GeWJ, ZhaoKM, WangXW, et al., 2017. iASPP is an antioxidative factor and drives cancer growth and drug resistance by competing with Nrf2 for Keap1 binding. Cancer Cell, 32(5):561-573.6.

[19]HarrisAL, 2002. Hypoxia—a key regulatory factor in tumour growth. Nat Rev Cancer, 2(1):38-47.

[20]HouP, ZhaoY, LiZ, et al., 2014. LincRNA-ROR induces epithelial-to-mesenchymal transition and contributes to breast cancer tumorigenesis and metastasis. Cell Death Dis, 5(6):e1287.

[21]HuZZ, DongN, LuD, et al., 2017. A positive feedback loop between ROS and Mxi1-0 promotes hypoxia-induced VEGF expression in human hepatocellular carcinoma cells. Cell Signal, 31:79-86.

[22]HumphriesBA, BuschhausJM, ChenYC, et al., 2019. Plasminogen activator inhibitor 1 (PAI1) promotes actin cytoskeleton reorganization and glycolytic metabolism in triple-negative breast cancer. Mol Cancer Res, 17(5):‍1142-1154.

[23]HurdTR, DeGennaroM, LehmannR, 2012. Redox regulation of cell migration and adhesion. Trends Cell Biol, 22(2):107-115.

[24]IvanM, KaelinWG, 2017. The EGLN-HIF O2-sensing system: multiple inputs and feedbacks. Mol Cell, 66(6):772-779.

[25]JinP, ShinSH, ChunYS, et al., 2018. Astrocyte-derived CCL20 reinforces HIF-1-mediated hypoxic responses in glioblastoma by stimulating the CCR6-NF-‍κB signaling pathway. Oncogene, 37(23):3070-3087.

[26]KeQD, CostaM, 2006. Hypoxia-inducible factor-1 (HIF-1). Mol Pharmacol, 70(5):1469-1480.

[27]KesslerJ, HahnelA, WichmannH, et al., 2010. HIF-1α inhibition by siRNA or chetomin in human malignant glioma cells: effects on hypoxic radioresistance and monitoring via CA9 expression. BMC Cancer, 10:605.

[28]KlimovaT, ChandelNS, 2008. Mitochondrial complex III regulates hypoxic activation of HIF. Cell Death Differ, 15(4):660-666.

[29]KunschC, MedfordRM, 1999. Oxidative stress as a regulator of gene expression in the vasculature. Circ Res, 85(8):753-766.

[30]LeiJJ, HuoXW, DuanWX, et al., 2014. α-Mangostin inhibits hypoxia-driven ROS-induced PSC activation and pancreatic cancer cell invasion. Cancer Lett, 347(1):129-138.

[31]LiSJ, WeiXH, ZhanXM, et al., 2020. Adipocyte-derived leptin promotes PAI-1-mediated breast cancer metastasis in a STAT3/miR-34a dependent manner. Cancers (Basel), 12(12):3864.

[32]LiX, WuP, TangYY, et al., 2020. Down-regulation of miR-181c-5p promotes epithelial-to-mesenchymal transition in laryngeal squamous cell carcinoma via targeting SERPINE1. Front Oncol, 10:544476.

[33]LiX, ZuoHW, ZhangL, et al., 2021. Validating HMMR expression and its prognostic significance in lung adenocarcinoma based on data mining and bioinformatics methods. Front Oncol, 11:720302.

[34]LiXD, DongP, WeiWS, et al., 2019. Overexpression of CEP72 promotes bladder urothelial carcinoma cell aggressiveness via epigenetic CREB-mediated induction of SERPINE1. Am J Pathol, 189(6):1284-1297.

[35]LiZW, HouPF, FanDM, et al., 2017. The degradation of EZH2 mediated by lncRNA ANCR attenuated the invasion and metastasis of breast cancer. Cell Death Differ, 24(1):59-71.

[36]LiuL, XiaoS, WangY, et al., 2022. Identification of a novel circular RNA circZNF652/miR-486-5p/SERPINE1 signaling cascade that regulates cancer aggressiveness in glioblastoma (GBM). Bioengineered, 13(1):1411-1423.

[37]LuW, KangYB, 2019. Epithelial-mesenchymal plasticity in cancer progression and metastasis. Dev Cell, 49(3):‍361-374.

[38]LukyanovaLD, KirovaYI, GermanovaEL, 2018. The role of succinate in regulation of immediate HIF-1α expression in hypoxia. Bull Exp Biol Med, 164(3):298-303.

[39]MittalM, GuXQ, PakO, et al., 2012. Hypoxia induces Kv channel current inhibition by increased NADPH oxidase-derived reactive oxygen species. Free Radic Biol Med, 52(6):1033-1042.

[40]Miyashita-IshiwataM, el SabehM, ReschkeLD, et al., 2022. Hypoxia induces proliferation via NOX4-mediated oxidative stress and TGF-β3 signaling in uterine leiomyoma cells. Free Radic Res, 56(2):163-172.

[41]MonteiroAR, HillR, PilkingtonGJ, et al., 2017. The role of hypoxia in glioblastoma invasion. Cells, 6(4):45.

[42]NiecknigH, TugS, ReyesBD, et al., 2012. Role of reactive oxygen species in the regulation of HIF-1 by prolyl hydroxylase 2 under mild hypoxia. Free Radic Res, 46(6):705-717.

[43]OlarA, AldapeKD, 2014. Using the molecular classification of glioblastoma to inform personalized treatment. J Pathol, 232(2):165-177.

[44]OmuroA, DeAngelisLM, 2013. Glioblastoma and other malignant gliomas: a clinical review. JAMA, 310(17):1842-1850.

[45]PengPH, LaiJCY, HsuKW, et al., 2020. Hypoxia-induced lncRNA RP11-390F4.3 promotes epithelial-mesenchymal transition (EMT) and metastasis through upregulating EMT regulators. Cancer Lett, 483:35-45.

[46]PerilloB, di DonatoM, PezoneA, et al., 2020. ROS in cancer therapy: the bright side of the moon. Exp Mol Med, 52(2):192-203.

[47]PolyakK, WeinbergRA, 2009. Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nat Rev Cancer, 9(4):265-273.

[48]QuinlanCL, OrrAL, PerevoshchikovaIV, et al., 2012. Mitochondrial complex II can generate reactive oxygen species at high rates in both the forward and reverse reactions. J Biol Chem, 287(32):27255-27264.

[49]SchieberM, ChandelNS, 2014. ROS function in redox signaling and oxidative stress. Curr Biol, 24(10):R453-R462.

[50]SchumackerPT, 2011. SIRT3 controls cancer metabolic reprogramming by regulating ROS and HIF. Cancer Cell, 19(3):299-300.

[51]SemenzaGL, 1998. Hypoxia-inducible factor 1: master regulator of O2 homeostasis. Curr Opin Genet Dev, 8(5):588-594.

[52]SemenzaGL, 2014. Oxygen sensing, hypoxia-inducible factors, and disease pathophysiology. Annu Rev Pathol Mech Dis, 9:47-71.

[53]SpencerNY, EngelhardtJF, 2014. The basic biology of redoxosomes in cytokine-mediated signal transduction and implications for disease-specific therapies. Biochemistry, 53(10):1551-1564.

[54]SrivastavaC, IrshadK, DikshitB, et al., 2018. FAT1 modulates EMT and stemness genes expression in hypoxic glioblastoma. Int J Cancer, 142(4):805-812.

[55]SubramanianA, TamayoP, MoothaVK, et al., 2005. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA, 102(43):15545-15550.

[56]SunB, YuL, XuC, et al., 2021. NAD(P)HX epimerase downregulation promotes tumor progression through ROS/HIF-1α signaling in hepatocellular carcinoma. Cancer Sci, 112(7):2753-2769.

[57]TakayamaY, HattoriN, HamadaH, et al., 2016. Inhibition of PAI-1 limits tumor angiogenesis regardless of angiogenic stimuli in malignant pleural mesothelioma. Cancer Res, 76(11):3285-3294.

[58]TangZH, ZhangZH, LinQQ, et al., 2021. HIF-1α/BNIP3-mediated autophagy contributes to the luteinization of granulosa cells during the formation of corpus luteum. Front Cell Dev Biol, 8:619924.

[59]TengF, ZhangJX, ChenY, et al., 2021. LncRNA NKX2-1-AS1 promotes tumor progression and angiogenesis via upregulation of SERPINE1 expression and activation of the VEGFR-2 signaling pathway in gastric cancer. Mol Oncol, 15(4):1234-1255.

[60]ThieryJP, AcloqueH, HuangRYJ, et al., 2009. Epithelial-mesenchymal transitions in development and disease. Cell, 139(5):871-890.

[61]TsaiJH, YangJ, 2013. Epithelial-mesenchymal plasticity in carcinoma metastasis. Genes Dev, 27(20):2192-2206.

[62]VordermarkD, 2005. Significance of hypoxia in malignant glioma. Re: Evanset al. Hypoxia is important in the biology and aggression of human glial brain tumors. Clin Cancer Res 2004;10:8177-84. Clin Cancer Res, 11(10):3966-3968.

[63]WangQ, LuWJ, YinT, et al., 2019. Calycosin suppresses TGF-‍β‍-induced epithelial-to-mesenchymal transition and migration by upregulating BATF2 to target PAI-1 via the Wnt and PI3K/Akt signaling pathways in colorectal cancer cells. J Exp Clin Cancer Res, 38:240.

[64]WangXW, BustosMA, ZhangXQ, et al., 2020. Downregulation of the ubiquitin-E3 ligase RNF123 promotes upregulation of the NF‍-‍κB1 target SerpinE1 in aggressive glioblastoma tumors. Cancers (Basel), 12(5):1081.

[65]WangZL, ShiYP, YingCT, et al., 2021. Hypoxia-induced PLOD1 overexpression contributes to the malignant phenotype of glioblastoma via NF‍-‍κB signaling. Oncogene, 40(8):1458-1475.

[66]WatsonJ, 2013. Oxidants, antioxidants and the current incurability of metastatic cancers. Open Biol, 3(1):120144.

[67]WeidemannA, JohnsonRS, 2008. Biology of HIF-1α. Cell Death Differ, 15(4):621-627.

[68]WillsonJA, ArientiS, SadikuP, et al., 2022. Neutrophil HIF-1α stabilization is augmented by mitochondrial ROS produced via the glycerol 3-phosphate shuttle. Blood, 139(2):281-286.

[69]WilsonWR, HayMP, 2011. Targeting hypoxia in cancer therapy. Nat Rev Cancer, 11(6):393-410.

[70]WuK, MaoYY, ChenQ, et al., 2021. Hypoxia-induced ROS promotes mitochondrial fission and cisplatin chemosensitivity via HIF-1α/Mff regulation in head and neck squamous cell carcinoma. Cell Oncol (Dordr), 44(5):‍1167-1181.

[71]XiaLM, MoP, HuangWJ, et al., 2012. The TNF-α/ROS/HIF-1-induced upregulation of FoxMI expression promotes HCC proliferation and resistance to apoptosis. Carcinogenesis, 33(11):2250-2259.

[72]XuBD, BaiZG, YinJ, et al., 2019. Global transcriptomic analysis identifies SERPINE1 as a prognostic biomarker associated with epithelial-to-mesenchymal transition in gastric cancer. PeerJ, 7:e7091.

[73]XuYQ, ChenWC, LiangJ, et al., 2021. The miR-1185-2-3p-GOLPH3L pathway promotes glucose metabolism in breast cancer by stabilizing p53-induced SERPINE1. J Exp Clin Cancer Res, 40:47.

[74]YangJ, AntinP, BerxG, et al., 2020. Guidelines and definitions for research on epithelial-mesenchymal transition. Nat Rev Mol Cell Biol, 21(6):341-352.

[75]YangJL, MaQ, ZhangMM, et al., 2021. LncRNA CYTOR drives L-OHP resistance and facilitates the epithelial-mesenchymal transition of colon carcinoma cells via modulating miR-378a-5p/SERPINE1. Cell Cycle, 20(14):1415-1430.

[76]YangY, ZhangGM, GuoFZ, et al., 2020. Mitochondrial UQCC3 modulates hypoxia adaptation by orchestrating OXPHOS and glycolysis in hepatocellular carcinoma. Cell Rep, 33(5):108340.

[77]YuLM, ZhangWH, HanXX, et al., 2019. Hypoxia-induced ROS contribute to myoblast pyroptosis during obstructive sleep apnea via the NF‍-‍κB/HIF‍-‍1αsignaling pathway. Oxid Med Cell Longev, 2019:4596368.

[78]ZhangW, ZhangYW, ZhouWS, et al., 2021. PlGF knockdown attenuates hypoxia-induced stimulation of cell proliferation and glycolysis of lung adenocarcinoma through inhibiting Wnt/β‍-catenin pathway. Cancer Cell Int, 21:18.

[79]ZhangYS, JinGS, ZhangJW, et al., 2018. Overexpression of STAT1 suppresses angiogenesis under hypoxia by regulating VEGF-A in human glioma cells. Biomed Pharmacother, 104:566-575.

[80]ZhaoQ, ZhangLW, HeQF, et al., 2023. Targeting TRMT5 suppresses hepatocellular carcinoma progression via inhibiting the HIF-1α pathways. J Zhejiang Univ-Sci B (Biomed & Biotechnol), 24(1):50-63.

[81]ZhengH, KangY, 2014. Multilayer control of the EMT master regulators. Oncogene, 33(14):1755-1763.

[82]ZongSQ, TangYF, LiW, et al., 2020. A Chinese herbal formula suppresses colorectal cancer migration and vasculogenic mimicry through ROS/HIF-1α/MMP2 pathway in hypoxic microenvironment. Front Pharmacol, 11:705.

Open peer comments: Debate/Discuss/Question/Opinion

<1>

Please provide your name, email address and a comment





Journal of Zhejiang University-SCIENCE, 38 Zheda Road, Hangzhou 310027, China
Tel: +86-571-87952783; E-mail: cjzhang@zju.edu.cn
Copyright © 2000 - 2024 Journal of Zhejiang University-SCIENCE